Sélection de la langue

Search

Sommaire du brevet 3073649 

Énoncé de désistement de responsabilité concernant l'information provenant de tiers

Une partie des informations de ce site Web a été fournie par des sources externes. Le gouvernement du Canada n'assume aucune responsabilité concernant la précision, l'actualité ou la fiabilité des informations fournies par les sources externes. Les utilisateurs qui désirent employer cette information devraient consulter directement la source des informations. Le contenu fourni par les sources externes n'est pas assujetti aux exigences sur les langues officielles, la protection des renseignements personnels et l'accessibilité.

Disponibilité de l'Abrégé et des Revendications

L'apparition de différences dans le texte et l'image des Revendications et de l'Abrégé dépend du moment auquel le document est publié. Les textes des Revendications et de l'Abrégé sont affichés :

  • lorsque la demande peut être examinée par le public;
  • lorsque le brevet est émis (délivrance).
(12) Demande de brevet: (11) CA 3073649
(54) Titre français: SCHEMAS DE TRAITEMENT
(54) Titre anglais: TREATMENT REGIMENS
Statut: Examen
Données bibliographiques
(51) Classification internationale des brevets (CIB):
  • A61K 31/7072 (2006.01)
  • A61P 35/00 (2006.01)
  • C07H 19/173 (2006.01)
  • C07H 19/20 (2006.01)
  • C12Q 01/48 (2006.01)
  • G01N 30/72 (2006.01)
  • G01N 33/48 (2006.01)
(72) Inventeurs :
  • GRIFFITH, HUGH (Royaume-Uni)
(73) Titulaires :
  • NUCANA PLC
(71) Demandeurs :
  • NUCANA PLC (Royaume-Uni)
(74) Agent: SMART & BIGGAR LP
(74) Co-agent:
(45) Délivré:
(86) Date de dépôt PCT: 2018-08-30
(87) Mise à la disponibilité du public: 2019-03-07
Requête d'examen: 2023-08-25
Licence disponible: S.O.
Cédé au domaine public: S.O.
(25) Langue des documents déposés: Anglais

Traité de coopération en matière de brevets (PCT): Oui
(86) Numéro de la demande PCT: PCT/GB2018/052455
(87) Numéro de publication internationale PCT: GB2018052455
(85) Entrée nationale: 2020-02-21

(30) Données de priorité de la demande:
Numéro de la demande Pays / territoire Date
1713916.3 (Royaume-Uni) 2017-08-30

Abrégés

Abrégé français

L'invention concerne le 5-fluoro-2'-désoxyuridine-5'-O-[1-naphtyl(benzoxy-L-alaninyl)]phosphate (NUC-3373), ou un sel pharmaceutiquement acceptable de celui-ci, destiné à être utilisé dans le traitement d'un cancer, en particulier par perfusion intraveineuse pendant une période continue allant jusqu'à 10 heures. L'invention concerne également des méthodes de traitement d'un cancer par administration de NUC-3373 à des sous-groupes particuliers de patients cancéreux. L'invention concerne en outre des procédés de sélection de patient pour un traitement par NUC-3373.


Abrégé anglais


The invention relates to 5-fluoro-2'-deoxyuridine-5'-O-[1-naphthyl (benzoxy-L-
alaninyl)] phosphate (NUC-3373), or a
pharmaceutically acceptable salt thereof, for use in the treatment of cancer,
in particular by intravenous infusion for a continuous period
of up to 10 hours. The invention also relates to methods of treating cancer by
administration of NUC-3373 to particular sub-groups of
cancer patient. The invention further relates to methods for selecting a
patient for treatment with NUC-3373.

Revendications

Note : Les revendications sont présentées dans la langue officielle dans laquelle elles ont été soumises.


32
Claims
1. 5-fluoro-2'-deoxyuridine-5'-O-[1-naphthyl (benzoxy-L-alaninyl)]
phosphate (NUC-
3373), or a pharmaceutically acceptable salt thereof, for use in the treatment
of cancer,
wherein the treatment is by administration of NUC-3373 over a period of up to
10 hours.
2. NUC-3373 for use according to claim 1, wherein the treatment is by
administration
of NUC-3373 over a period of up to 5 hours.
3. NUC-3373 for use according to claim 1 or claim 2, wherein the treatment
is by
administration of NUC-3373 over a period of up to 2 hours.
4. NUC-3373 for use according to claim 1, wherein the treatment is by
administration
of NUC-3373 over a period of between 1 and 2 hours, 2 and 4 hours or 1 and 6
hours.
5. NUC-3373 for use according to any preceding claim, wherein the
administration is
by means of continuous infusion.
6. NUC-3373 for use according to claim 5, wherein the infusion is by
intravenous
infusion.
7. NUC-3373 for use according to any of the preceding claims, wherein the
treatment
is by administration of NUC-3373 by means of or includes a bolus
administration.
8. NUC-3373 for use according to any preceding claim, wherein the cancer is
selected from the group consisting of: pancreatic cancer, breast cancer,
ovarian cancer,
bladder cancer, other urothelial cancers, gastrointestinal cancer (also known
as cancer of
the digestive tract), liver cancer, lung cancer, biliary cancer, prostate
cancer,
cholangiocarcinoma, renal cancer, neuroendocrine cancer, sarcoma, lymphoma,
leukemia, cervical cancer, thymic cancer, a cancer of an unknown primary
origin,
mesothelioma, adrenal cancer, cancer of the uterus, cancer of the fallopian
tube,
peritoneal cancer, endometrial cancer, testicular cancer, head and neck
cancer, the
central nervous system cancer, basal cell carcinoma, Bowens disease, other
skin cancers
(such as malignant melanoma, merckel cell tumour and rare appendage tumours),
ocular
surface squamous neoplasia and germ cell tumours.
9. NUC-3373 for use according to claim 8, wherein the cancer is a
gastrointestinal
cancer selected from the group consisting of: oesophageal cancer, gastric
cancer,
stomach cancer, bowel cancer, small intestine cancer, colon cancer, appendix
mucinous,
goblet cell carcinoid, liver cancer, biliary cancer, gallbladder cancer, anal
cancer and
rectal cancer.

33
10. NUC-3373 for use according to any preceding claim, wherein the patient
with the
cancer also suffers from hand-foot syndrome.
11. NUC-3373 for use according to claim 10, wherein the patient has
developed hand-
foot syndrome from a previous treatment regimen with a drug other than NUC-
3373.
12. NUC-3373 for use according to claim 11, wherein the patient has
developed hand-
foot syndrome when being treated with 5FU, capecitabine or tegafur.
13. 5-fluoro-2'-deoxyuridine-5'-O-[1-naphthyl (benzoxy-L-alaninyl)]
phosphate (NUC-
3373) or a pharmaceutically acceptable salt thereof, for use in the treatment
of cancer in a
subject that suffers from hand-foot syndrome.
14. NUC-3373 for use according to claim 14, wherein the subject has
developed hand-
foot syndrome following treatment with a fluoropyrimidine such as 5FU,
capecitabine or
tegafur.
15. NUC-3373 for use according to claim 14 or 15, wherein the cancer is
selected from
the group consisting of: pancreatic cancer, breast cancer, ovarian cancer,
bladder cancer,
other urothelial cancers, gastrointestinal cancer (also known as cancer of the
digestive
tract), liver cancer, lung cancer, biliary cancer, prostate cancer,
cholangiocarcinoma, renal
cancer, neuroendocrine cancer, sarcoma, lymphoma, leukemia, cervical cancer,
thymic
cancer, a cancer of an unknown primary origin, mesothelioma, adrenal cancer,
cancer of
the uterus, cancer of the fallopian tube, peritoneal cancer, endometrial
cancer, testicular
cancer, head and neck cancer, the central nervous system cancer, basal cell
carcinoma,
Bowens disease, other skin cancers (such as malignant melanoma, merckel cell
tumour
and rare appendage tumours), ocular surface squamous neoplasia and germ cell
tumours.
16. NUC-3373 for use according to any of claims 14 to 16, wherein the
cancer is
gastrointestinal cancer and is selected from the group consisting of:
oesophageal cancer,
gastric cancer, stomach cancer, bowel cancer, small intestine cancer, colon
cancer,
appendix mucinous, goblet cell carcinoid, liver cancer, biliary cancer,
gallbladder cancer,
anal cancer and rectal cancer.
17. A method of selecting a subject with cancer for treatment with 5-fluoro-
2'-
deoxyuridine-5'-O-[1-naphthyl (benzoxy-L-alaninyl)] phosphate (NUC-3373), or a
pharmaceutically acceptable salt thereof, the method comprising determining
whether the
subject has hand-foot syndrome, wherein if the subject has hand-foot syndrome,
the
subject is selected for treatment with 5-fluoro-2'-deoxyuridine-5'-O-[1-
naphthyl (benzoxy-
L-alaninyl)] phosphate (NUC-3373).

34
18. The method according to claim 18, wherein the patient developed hand-
foot
syndrome whilst being treated with a fluoropyrimidine, such as 5FU or
capecitabine.
19. 5-fluoro-2'-deoxyuridine-5'-O-[1-naphthyl (benzoxy-L-alaninyl)]
phosphate (NUC-
3373), or a pharmaceutically acceptable salt thereof, for use in the treatment
of cancer in
subjects that are deficient or partially deficient in dihydropyrimidine
dehydrogenase (DPD).
20. NUC-3373 for use according to claim 20, wherein the cancer is selected
from the
group consisting of: pancreatic cancer, breast cancer, ovarian cancer, bladder
cancer,
other urothelial cancers, gastrointestinal cancer (also known as cancer of the
digestive
tract), liver cancer, lung cancer, biliary cancer, prostate cancer,
cholangiocarcinoma, renal
cancer, neuroendocrine cancer, sarcoma, lymphoma, leukemia, cervical cancer,
thymic
cancer, a cancer of an unknown primary origin, mesothelioma, adrenal cancer,
cancer of
the uterus, cancer of the fallopian tube, peritoneal cancer, endometrial
cancer, testicular
cancer, head and neck cancer, the central nervous system cancer, basal cell
carcinoma,
Bowens disease, other skin cancers (such as malignant melanoma, merckel cell
tumour
and rare appendage tumours), ocular surface squamous neoplasia and germ cell
tumours.
21. NUC-3373 for use according to claim 20 or 21, wherein the cancer is
gastrointestinal cancer and is selected from the group consisting of:
oesophageal cancer,
gastric cancer, stomach cancer, bowel cancer, small intestine cancer, colon
cancer,
appendix mucinous, goblet cell carcinoid, liver cancer, biliary cancer,
gallbladder cancer,
anal cancer and rectal cancer.
22. NUC-3373 for use according to any one of claims 20 to 22, wherein the
subject
has a genetic mutation selected from IVS14+1G>A mutation in intron 14 coupled
with
exon 14 deletion (known as DPYD*2A), 496A>G in exon 6; 2846A>T in exon 22; and
T1679G (DPYD*13) in exon 13.
23. NUC-3373 for use according to claim 23, wherein the subject has the
IV514+1G>A DPYD variant (DPYD*2A) mutation.
24. NUC-3373 for use according to any one of claims 20 to 22, wherein the
subject
has previously exhibited intolerance for 5FU or capecitabine or has a family
history of
intolerance for 5FU or capecitabine.
25. A method of assessing effectiveness of an anti-cancer therapy, the
method
comprising: assaying a sample of peripheral blood mononuclear cells (PBMCs) or
cancer
cells from a subject receiving an anti-cancer therapy to determine the level
of intracellular
deoxythymidine monophosphate (dTMP) within the PBMCs or cancer cells, wherein
a

35
reduction in the level of intracellular dTMP within the PBMCs or cancer cells
indicates that
the anti-cancer therapy is effective.
26. A method according to claim 26, wherein the subject is receiving anti-
cancer
treatment using NUC-3373.
27. A method according to claim 26 or claim 27, wherein the level of
intracellular
dTMP within the PBMCs or cancer cells is compared to a suitable control value.
28. A method according to any of claims 26 to 28, wherein the reduction is
a reduction
of at least 25%.
29. A method according to claim 29, wherein the reduction is substantially
a complete
reduction of intracellular dTMP.
30. A method of assessing effectiveness of an anti-cancer therapy, the
method
comprising: assaying a sample of peripheral blood mononuclear cells (PBMCs) or
cancer
cells from a subject receiving an anti-cancer therapy to determine the level
of intracellular
thymidylate synthase (TS) within the PBMCs or cancer cells, wherein a
reduction in the
level of intracellular TS within the PBMCs or cancer cells indicates that the
anti-cancer
therapy is effective.
31. A method according to claim 31, wherein the subject is receiving anti-
cancer
treatment using NUC-3373.
32. A method according to claim 31 or claim 32, wherein the level of
intracellular TS
within the PBMCs or cancer cells is compared to a suitable control value.
33. A method according to any of claims 31 to 33, wherein the reduction is
a reduction
of at least 25%.
34. A method according to claim 34 wherein the reduction is substantially a
complete
reduction of intracellular TS.

Description

Note : Les descriptions sont présentées dans la langue officielle dans laquelle elles ont été soumises.


CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
1
TREATMENT REGIMENS
[0001] The present invention relates to 5-fluoro-2'-deoxyuridine-5'-041-
naphthyl
(benzoxy-L-alaninyI)] phosphate (NUC-3373), or a pharmaceutically acceptable
salt
thereof, for use in the treatment of cancer. The invention also relates to
methods of
treating cancer by administration of NUC-3373. The invention further relates
to methods
of assessing effectiveness of anti-cancer treatments.
The invention further relates to
patient groups that would particularly benefit from treatment with NUC-3373.
BACKGROUND
NUC-3373
[0002] Protides are masked phosphate derivatives of nucleosides. They have
been
shown to be particularly potent therapeutic agents in the fields of both
antivirals and
oncology. Protides appear to avoid many of the inherent and acquired
resistance
mechanisms which limit the utility of the parent nucleosides.
[0003] A ProTide adaptation of the nucleoside analogue, 5FUDR, 5-fluoro-2'-
deoxyuridine-5'-0[l-naphthyl (benzoxy-L-alaninyI)] phosphate (NUC-3373) 1 and
a range
of related compounds have shown activity in vitro against a range of cancer
models, in
many cases and in particular for NUC-3373 that activity was outstanding and
far superior
to the results obtained with 5-fluorouracil. The addition of the protide
phosphoramidate
moiety to the 5-fluorouracil/FUDR molecule confers the specific advantages of
delivering
the key activated form of the agent (FUDR monophosphate) into the tumour
cells. Non
clinical studies have demonstrated that NUC-3373 overcomes the key cancer cell
resistance mechanisms associated with 5FU and its oral pro-drug capecitabine,
generating high intracellular levels of the active FUDR monophosphate
metabolite,
resulting in a much greater inhibition of tumour cell growth. Furthermore, in
formal dog
toxicology studies, NUC-3373 is significantly better tolerated than 5FU
(see
W02012/117246; McGuigan et al.; Phosphoramidate ProTides of the anticancer
agent
FUDR successfully deliver the preformed bioactive monophosphate in cells and
confer
advantage over the parent nucleoside; J. Med. Chem.; 2011, 54, 7247-7258; and
Vande
Voorde et al.; The cytostatic activity of NUC-3073, a phosphoramidate prodrug
of 5-fluoro-
2'-deoxyuridine, is independent of activation by thymidine kinase and
insensitive to
degradation by phosphorolytic enzymes; Biochem. Pharmacol.; 2011, 82, 441-
452).

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
2
0
FII
)L
0 NH
O-P-0
N 0
401 I
NH
OH
0 1
[0004] NUC-3373 1 is typically prepared as a mixture of two diastereoisomers,
epimeric
at the phosphate centre (the S-epimer and the R-epimer).
[0005] The therapeutic effect of 5-fluorouracil is through the formation of
nucleotides that
block normal nucleic acid formation. This is balanced by catabolism by
dihydropyrimidine
dehydrogenase (DPD) in the liver. Cancer patients that are dihydropyrimidine
dehydrogenase deficient (DPD) or partially deficient are unable to degrade 5FU
and other
chemotherapeutic agents; most degradation occurs in the liver (DeLeve. Drug-
Induced
Liver Disease (Third Edition) 2013, pages 541-567). More than 85% of 5-
fluorouracil is
broken down by DPD; therefore, DPD activity is a major determinant of 5-
fluorouracil
activity and toxicity. DPD enzyme activity follows a Gaussian distribution
with up to six-fold
inter-individual variation. In addition to the normal variation of DPD
activity, there are -also
imitations in the DPYD gene that can lead to DPD deficiency, which occurs in
less than
3% of the population. Low DPD levels and DPD deficiency reduce 5-fluorouracil
clearance
and can lead to severe or life-threatenino toxicity.
[0006] In a clinical study it was observed that 55% of patients with a
decreased DPD
activity suffered from grade IV neutropenia compared with 13% of patients with
a normal
DPD activity (P = 0.01). Furthermore, the onset of toxicity occurred, on
average, twice as
fast in patients with low DPD activity as compared with patients with a normal
DPD activity
(10.0 +1- 7.6 versus 19.1 +1- 15.3 days; P < 0.05) (van Kullenburg et al.,
Clin Cancer res.
6(12):4705-12, 2000).
To date, 39 different mutations and polymorphisms have been identified in
DPYD. The
IVS14+1G>A mutation proved to be the most common one and was detected in 24-
28%
of all patients suffering from severe 5FU toxicity. Thus, a deficiency of DPD
appears to be
an important pharmacogenetic syndrome (van Kullenberg. Eur. J. Cancer.
40(7):939-50,
2004).
[0007] The reduced liver degradation of, for example, 5FU leads to prolonged
exposure
to the agent which can lead to profound toxicity, including mucositis,
diarrhoea,
granulocytopenia, neuropathy and death (Jin Ho Baek et al. Korean J
International

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
3
Medicine. 21:43-45, 2006). Conversely, patients that are not DPD deficient
(e.g. normal
DPD) are able to breakdown 5FU (and certain other chemotherapeutic agents) in
the liver,
however, 5FU is broken down to potentially toxic metabolites such as a-fluoro-
8-alanine
(FBAL) and dihydrofluorouracil (dhFU) presence of which is associated with
hand-foot
syndrome (also known as chemotherapy-induced acral erythema or palmar-plantar
erythrodysesthesia, palmoplantar erythrodysesthesia) in 30-60% of patients
(Kruger et al.
Acta Oncologica 1-8, 2015; Chiara et al. Eur J Cancer. 33:967-969, 1997).
Although not
life threatening this can be debilitating.
[0008] There is therefore a need to identify treatments for cancer with a
better safety
profile (fewer side effects) than observed with existing therapies, such as
5FU, and
capecitabine or Tegafur (prodrugs of 5FU), so that patients can either be
treated with
these treatments (e.g. agents) first line, or can be switched to these agents
when the
patient develops a side effect such as hand-foot syndrome, or is identified as
a patient
likely to develop such a side effect and/or less toxic side effects.
.. [0009] It is an aim of this invention to provide a therapy regime for
treating cancer. It is
also an aim of the present invention to provide methods for treating cancer
patients who
have DPD deficiency or partial DPD deficiency or who develop hand-foot
syndrome. It is
also an aim of this invention to provide methods for determining the
effectiveness of a
therapy for treating cancer. It is also an aim of this invention to provide
methods for
determining whether a subject's cancer is DPD deficient or partially deficient
and providing
a therapy for treating such a cancer.
[0010] It is an aim of this invention to provide a therapy that is more
effective and/ or
less toxic/safer than existing treatments.
[0011] Certain embodiments of this invention satisfy some or all of the above
aims.
BRIEF SUMMARY OF THE DISCLOSURE
[0012] In a first aspect, the invention provides 5-fluoro-2'-deoxyuridine-5'-
0[l-naphthyl
(benzoxy-L-alaninyI)] phosphate (NUC-3373), or a pharmaceutically acceptable
salt
thereof, for use in the treatment of cancer, wherein the treatment is by
administration of
NUC-3373 over a period of up to 10 hours, such as between 1 and 6 hours.
[0013] In a second aspect the invention provides a method of treating cancer,
the
method comprising administering 5-fluoro-2'-deoxyuridine-5'-0[l-naphthyl
(benzoxy-L-
alaniny1)] phosphate (NUC-3373), or a pharmaceutically acceptable salt
thereof, to a
subject in need of such treatment over a period of up to 10 hours.

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
4
[0014] In a third aspect the invention provides the invention provides the use
of 5-fluoro-
2'-deoxyuridine-5'-041-naphthyl (benzoxy-L-alaninyI)] phosphate (NUC-3373), or
a
pharmaceutically acceptable salt thereof, in the manufacture of a medicament
for use in
the treatment of cancer, wherein the treatment is by administration of the
medicament
comprising NUC-3373 over a period of up to 10 hours.
[0015] In a fourth aspect, the invention provides a method of assessing
effectiveness of
an anti-cancer therapy, the method comprising:
assaying a sample of peripheral blood mononuclear cells (PBMCs) or cancer
cells from a
subject receiving an anti-cancer therapy to determine the level of
intracellular
deoxythymidine monophosphate (dTMP) within the PBMCs or cancer cells,
wherein a reduction in the level of intracellular dTMP within the PBMCs or
cancer cells
indicates that the anti-cancer therapy is effective.
[0016] In a fifth aspect, the invention provides a method of assessing
effectiveness of an
anti-cancer therapy, the method comprising:
assaying a sample of peripheral blood mononuclear cells (PBMCs) or cancer
cells from a
subject receiving an anti-cancer therapy to determine the level of
intracellular thymidylate
synthase (TS) within the PBMCs or cancer cells,
wherein a reduction in the level of intracellular TS within the PBMCs or
cancer cells
indicates that the anti-cancer therapy is effective.
On exposure to 5FU the levels of TS may increase and may contribute to
resistance.
Because treatment with NUC-3373 results in greater levels of the inhibitor
FUDRMP it is
predicted that treatment with NUC-3373 will be effective even in patients with
increased
levels of TS, that have typically arisen due to resistance mechanisms
following treatment
with agents such as 5FU.
[0017] In a sixth aspect, the invention provides a method of assessing
effectiveness of
an anti-cancer therapy, the method comprising:
assaying a sample of peripheral blood mononuclear cells (PBMCs) or cancer
cells from a
subject receiving an anti-cancer therapy to determine the level of
intracellular
deoxyuridine monophosphate (dUMP) within the PBMCs or cancer cells,
wherein an increase in the level of intracellular dUMP within the PBMCs or
cancer cells
indicates that the anti-cancer therapy is effective.

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
[0018] It will be appreciated that suitable cancer cells for use in the
methods of the
fourth to sixth aspects of the invention may be selected with reference to the
subject's
cancer. Peripheral blood mononuclear cells (PBMCs) are blood cells with
nuclei, such as
monocytes, lymphocytes, and macrophages.
5 In a seventh aspect, the invention provides 5-fluoro-2'-deoxyuridine-5'-
0[l-naphthyl
(benzoxy-L-alaninyl)] phosphate (NUC-3373), or a pharmaceutically acceptable
salt
thereof, for use in the treatment of cancer by increasing intracellular levels
of dUMP in
treated cancer cells, and thereby causing cancer cell death. Without wishing
to be bound
by theory, in this scenario it may be that the cancer cell is killed due to a
reduction in the
efficacy of cellular DNA repair mechanism or due to cell stress.
[0019] In an eighth aspect, the invention provides 5-fluoro-2'-deoxyuridine-5'-
041-
naphthyl (benzoxy-L-alaninyl)] phosphate (NUC-3373), or a pharmaceutically
acceptable
salt thereof, for use in the treatment of cancer in subjects that are
deficient or partially
deficient in dihydropyrimidine dehydrogenase (DPD).
In an embodiment, the eighth aspect of the invention provides 5-fluoro-2'-
deoxyuridine-5'-
041-naphthyl (benzoxy-L-alaninyl)] phosphate (NUC-3373) or a pharmaceutically
acceptable salt thereof, for use in treatment of a cancer in a subject that
has been
identified as being deficient or partially deficient in DPD.
In a ninth aspect, the invention provides a method of treating cancer, the
method
comprising administering 5-fluoro-2'-deoxyuridine-5'-041-naphthyl (benzoxy-L-
alaninyl)]
phosphate (NUC-3373), or a pharmaceutically acceptable salt thereof, to a
subject in
need of such treatment wherein the subject is deficient or partially deficient
in
dihydropyrimidine dehydrogenase (DPD). The subject may be identified as
deficient or
partially deficient in DPD by any suitable means, including those disclosed
herein.
[0020] NUC-3373 for use in accordance with any of the seventh to tenth aspects
may be
employed in accordance with any of the methods of treatment or medical uses,
in their
various aspects or embodiments, described herein.
[0021] In a tenth aspect, the invention provides a method of selecting a
subject for
treatment with 5-fluoro-2'-deoxyuridine-5'-041-naphthyl (benzoxy-L-alaninyl)]
phosphate
(NUC-3373), or a pharmaceutically acceptable salt thereof, the method
comprising
determining whether the subject's liver cells, PBMCs or a cancer cell are
deficient or
partially deficient in DPD, wherein if the subject's liver cells, PBMCs or
cancer cell are
DPD deficient the subject is selected for treatment with 5-fluoro-2'-
deoxyuridine-5'-041-
naphthyl (benzoxy-L-alaninyl)] phosphate (NUC-3373).

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
6
[0022] In an embodiment, the method of determining whether the subject's liver
cells,
PBMCs or a cancer cell are deficient or partially deficient in DPD is carried
out by
determining the amount or activity of DPD in a sample of PBMCs or cancer cells
from the
subject as compared to a reference value, wherein if the subject's PBMCs or
cancer cells
are deficient or partially deficient in DPD, the subject is selected for
treatment with 5-
fluoro-2'-deoxyuridine-5'-041-naphthyl (benzoxy-L-alaninyI)] phosphate (NUC-
3373).
[0023] In an eleventh aspect, the invention provides a method for determining
whether a
patient suffering with cancer is likely to benefit from or be responsive to
treatment with 5-
fluoro-2'-deoxyuridine-5'-041-naphthyl (benzoxy-L-alaninyI)] phosphate (NUC-
3373), or a
.. pharmaceutically acceptable salt thereof, the method comprising determining
the amount
of activity of DPD protein expressed in a cancer cell isolated from the
patient, wherein if
the patient's cancer cell expresses reduced amount or DPD relative to a
reference value,
the patient is likely to benefit from or be responsive to treatment with 5-
fluoro-2'-
deoxyuridine-5'-041-naphthyl (benzoxy-L-alaninyI)] phosphate (NUC-3373), or a
pharmaceutically acceptable salt thereof. In an embodiment, determination of
whether a
patient is deficient or partially deficient in DPD is carried out on a cancer
cell previously
isolated from the patient suffering with cancer.
[0024] In a twelfth aspect, the invention provides use of 5-fluoro-2'-
deoxyuridine-5'-041-
naphthyl (benzoxy-L-alaninyI)] phosphate (NUC-3373) or a pharmaceutically
acceptable
salt thereof, in a method of treating a subject with cancer which is deficient
or partially
deficient in DPD.
[0025] In a thirteenth aspect, the invention provides use of 5-fluoro-2'-
deoxyuridine-5'-0-
[1-naphthyl (benzoxy-L-alaninyI)] phosphate (NUC-3373) or a pharmaceutically
acceptable salt thereof, in the manufacture of a medicament for use in a
method of
treating a subject with cancer, the method comprising:
(i) determining whether the subject's cancer is deficient or partially
deficient in DPD and
(ii) administering to a subject whose cancer has been identified as
being deficient or partially deficient in DPD a pharmaceutically effective
amount of 5-
fluoro-2'-deoxyuridine-5'-041-naphthyl (benzoxy-L-alaninyI)] phosphate (NUC-
3373).
[0026] In a particular embodiment, the subject's cancer is determined to be
deficient or
partially deficient in DPD by testing a cancer cell containing sample
previously isolated
from the subject for DPD activity, the presence of DPD protein, or surrogate
thereof. As
used herein the term patient and subject are used interchangeably.

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
7
[0027] By way of example, a suitable surrogate may include mRNA, DPD
degradation
products, or the ratio of dihydrouracil to uracil ratio in plasma.
[0028] In a fourteenth aspect, the invention provides 5-fluoro-2'-deoxyuridine-
5'-041-
naphthyl (benzoxy-L-alaninyl)] phosphate (NUC-3373) or a pharmaceutically
acceptable
salt thereof, for use in the treatment of cancer in a subject that is at risk
of developing or
develops (has developed) hand-foot syndrome when being treated for their
cancer by an
agent other than NUC-3373. In particular embodiments, the patient is at risk
of developing
or develops hand-foot syndrome when being treated with or following treatment
with 5FU
or capecitabine. In a further aspect there is provided 5-fluoro-2'-
deoxyuridine-5'-041-
naphthyl (benzoxy-L-alaninyl)] phosphate (NUC-3373) or a pharmaceutically
acceptable
salt thereof, for use in the treatment of cancer in a subject that suffers
from hand-foot
syndrome. In a particular embodiment, the subject has developed hand-foot
syndrome
following treatment with a drug other than NUC-3373. In particular
embodiments, the 'drug
other than NUC-3373' is a fluoropyrimidine such as 5FU, capecitabine or
tegafur.
[0029] It is known that 30-60% of patients treated with 5FU develop skin
anomalies,
such as hand-foot syndrome. Such patients typically express normal levels of
DPD. Such
potentially debilitating side effects are likely due to the build-up of toxic
byproducts such
as a-fluoro-8-alanine (FBAL) and dihydrofluorouracil (dhFU) when treated with
anti-cancer
agents such as 5FU and capecitabine (a prodrug of 5FU). FBAL and dhFU, were
only
detected at very low levels or were undetectable, respectively, following NUC-
3373
administration at the doses studied and so NUC-3373 treatment is not expected
to result
in the side effects associated with FBAL and dhFU, such as hand-foot syndrome.
This
means that patients that develop hand-foot syndrome (or are at risk of
developing this
syndrome) when being treated for their cancer, such as when treated with 5FU
or
capecitabine, would benefit from switching to NUC-3373 treatment.
[0030] In an embodiment, the fourteenth aspect of the invention provides 5-
fluoro-2'-
deoxyuridine-5'-041-naphthyl (benzoxy-L-alaninyl)] phosphate (NUC-3373) or a
pharmaceutically acceptable salt thereof, for use in the treatment of a cancer
in a subject
that has developed hand-foot syndrome following treatment with 5FU or
capecitabine.
[0031] In a fifteenth aspect, the invention provides a method of treating
cancer, the
method comprising administering 5-fluoro-2'-deoxyuridine-5-0[l-naphthyl
(benzoxy-L-
alaninyl)] phosphate (NUC-3373), or a pharmaceutically acceptable salt
thereof, to a
cancer subject with hand-foot syndrome. In particular embodiments, the patient
has
developed hand-foot syndrome by virtue of being treated with 5FU or
capecitabine. In a
particular embodiment, the patient's cancer treatment is switched to NUC-3373.

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
8
[0032] In a sixteenth aspect, the invention provides use of 5-fluoro-2'-
deoxyuridine-5'-0-
[1-naphthyl (benzoxy-L-alaninyI)] phosphate (NUC-3373) or a pharmaceutically
acceptable salt thereof, in the manufacture of a medicament for use in a
method of
treating a subject with cancer, the method comprising:
(i) determining whether the cancer subject has hand-foot syndrome and
(ii) administering to the cancer subject with hand-foot syndrome a
pharmaceutically
effective amount of 5-fluoro-2'-deoxyuridine-5'-0[1-naphthyl (benzoxy-L-
alaninyI)]
phosphate (NUC-3373).
[0033] In a seventeenth aspect, the invention provides a method of selecting a
subject
.. with cancer for treatment with 5-fluoro-2'-deoxyuridine-5'-0[1-naphthyl
(benzoxy-L-
alaniny1)] phosphate (NUC-3373), or a pharmaceutically acceptable salt
thereof, the
method comprising determining whether the subject has hand-foot syndrome,
wherein if
the subject has hand-foot syndrome, the subject is selected for treatment with
5-fluoro-2'-
deoxyuridine-5'-041-naphthyl (benzoxy-L-alaninyI)] phosphate (NUC-3373).
[0034] NUC-3373 for use in accordance with any of the twelfth to seventeenth
aspects
may be employed in accordance with any of the methods of treatment or medical
uses, in
their various aspects or embodiments, described herein.
[0035] In an eighteenth aspect, the invention provides a method for
determining whether
delivery of an active anti-cancer agent into a cell has been achieved, the
method
comprising:
assaying a sample of peripheral blood mononuclear cells (PBMCs) or cancer
cells from a
subject receiving an anti-cancer therapy to determine the level of
intracellular
deoxyuridine monophosphate (dUMP) within the PBMCs or cancer cells, wherein an
increase in the level of intracellular dUMP within the PBMCs or cancer cells
indicates that
delivery of the active anti-cancer agent into the cell has been achieved. In a
particular
embodiment the active agent is NUC-3373.
BRIEF DESCRIPTION OF THE FIGURES
[0036] Embodiments of the invention are further described hereinafter with
reference to
the accompanying drawings, in which:
Figure 1 shows the Cm, and AUC for NUC-3373 in the blood plasma.
Figure 2 shows the Cm, and AUC for intracellular FUDR monophosphate.

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
9
DETAILED DESCRIPTION
The first to third aspects of the present invention are based upon the
inventors' surprising
finding that NUC-3373 is retained in the circulation after administration for
a period that is
much longer than for 5-fluorouracil (5FU), the "parent" compound from which
NUC-3373 is
derived. As explained further in the Examples, the inventors have found that
NUC-3373
has a plasma half-life of approximately 9.4 hours, as compared to a half-life
of only 8-14
minutes for 5FU. This difference means that therapeutically effective levels
of NUC-3373
are maintained for much longer after administration of this agent than after
continual 46-
48 hour administration of 5FU. As a consequence, effective anti-cancer
treatment with
NUC-3373 can make use of administration periods of 5/10 hours, or less, such
as 3 or 4
hours, and even as little as 1 or 2 hours. This is in marked contrast to
current treatment
protocols with 5FU, the current standard of care for the treatment of many
cancers. Here,
the shorter half-life of 5FU requires the agent to be optimally administered
to a patient
over long periods, in order to achieve therapeutic activity. Treatment via
constant infusion
of 5FU over a period of 46-48 hours is commonplace, highlighting the short
half-life (8-14
minutes) of 5FU. Sometimes this infusion is preceded by administration of a
bolus dose
(short infusion) of 5FU. Constant infusion refers to the administration of a
fluid to a blood
vessel, usually over a prolonged period of time.
[0037] It will be appreciated that the ability of the medical uses and methods
of
treatment of the invention to achieve therapeutically effective levels of NUC-
3373 in a
subject using much shorter periods of infusion offer many notable advantages.
One major
benefit of the medical uses and methods of treatment of the invention lies in
the
decreased disruption and invasiveness suffered by the recipient. Prior
treatments' need
for prolonged infusion periods, in the region of 46 - 48 hours, significantly
decrease the
quality of life experienced by the recipient. It will be appreciated that a
reduced
administration period results in a reduction in the time spend in hospital,
and thus a
likelihood of an increase in quality of life experienced by the recipient. A
reduced
administration period also results in decreased healthcare costs.
The methods of assessing effectiveness of an anti-cancer therapy provided by
the fourth
fifth, and sixth aspects of the invention arise from the inventors' surprising
finding that
treatment with NUC-3373 is able to deplete dTMP and TS levels and/or activity
in the cells
of subjects to whom it is administered and because dUMP is not converted to
dTMP,
treatment with NUC-3373 leads to an increased accumulation of intracellular
dUMP. This
is a new finding, and one that provides not only an indication of the way in
which NUC-
3373 may be achieving its therapeutic effect, but also provides a way in which
the

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
effectiveness of treatment may be monitored. Depletion of intracellular dTMP
or TS levels
or activity in PBMCs is not observed in patients receiving 5FU. Once again,
this provides
an indication of the different, and increased, effectiveness of NUC-3373 as
compared to
its parent compound. TS catalyses the
conversion of deoxyuridine
5
monophosphate (dUMP) to deoxythymidine monophosphate (dTMP). dTMP is
subsequently phosphorylated to produce dTTP, a vital precursor for DNA
replication and
repair (VVilson et al., Nat Rev Olin Oncol. 11(5):282-98, 2014). Therefore, a
reduction in
the level of intracellular TS or inhibition of TS results in increased levels
of dUMP in the
cell which results in DNA damage.
10 [0038] It is this identification of a new mode of action of NUC-3373
that underlies the
medical use of the seventh aspect of the invention. Recognition of this new
mode of
action, in which treatment with NUC-3373 increases intracellular accumulation
of dUMP in
cancer cells, thus causing the cells' death, enables novel uses of this
compound in new
clinical applications.
In an embodiment of the seventh aspect of the invention, NUC-
3373 may act to kill cancer cells by increasing levels of dUMP in the cells
which results in
DNA damage. The DNA damage then causes death of the cells, thereby bringing
about
effective treatment of cancer.
[0039] The medical uses and methods of treatment of the eighth, ninth, twelfth
and
thirteenth aspects of the present invention, and the patient selection and
determination
methods of the tenth and eleventh aspects of the invention, all relate to
cancers that are
deficient or partially deficient in DPD. These aspects are based upon the
inventors'
surprising finding that the toxic byproducts of 5FU administration, a-fluoro-8-
alanine
(FBAL) and dihydrofluorouracil (dhFU), were only detected at very low levels
or were
undetectable following NUC-3373 administration at the doses studied. Such
byproducts
are produced by 5FU and also produced when capecitabine, a prodrug of 5FU are
administered, at clinically relevant amounts (leading to side effects such as
hand-foot
syndrome).
[0040] The medical uses and methods of treatment of the fourteenth to
seventeenth
aspects of the invention, all relate to cancer patients who develop side
effects, such as
hand-foot syndrome, when on chemotherapeutic agents, such as 5FU or
capecitabine,
due to the build-up of toxic metabolites (e.g. dhFU, FBAL) whose presence is
associated
with hand-foot syndrome. Again, these aspects are based upon the inventors'
surprising
finding that the toxic byproducts of 5FU administration, a-fluoro-8-alanine
(FBAL) and
dihydrofluorouracil (dhFU), were only detected at very low levels or were
undetectable
following NUC-3373 administration at the doses studied.

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
11
[0041] Since DPD is involved in pyrimidine degradation, subjects that are DPD
deficient
or partially deficient are unable to metabolise 5FU or capecitabine.
[0042] Patients with DPD deficiency who are treated with 5FU or capecitabine
are at
significantly increased risk of developing severe (grade III/IV) and
potentially fatal
.. neutropenia, mucositis and diarrhoea. DPD deficiency effects 5% of the
population and a
further 3-5% of the population are partially DPD deficient. The reduction in
the amount of
these by products (e.g. FBAL and dhFU) in patients treated with NUC-3373
therefore
would suggest that DPD deficient and partially deficient patients can be
treated with NUC-
3373 whereas they could not be treated with 5FU or capecitabine. Therefore,
NUC-3373
provides a lower risk treatment option for DPD deficient and partially
deficient patients
over 5FU or capecitabine.
[0043] Patients treated with 5FU or capecitabine that are not deficient in
DPD, often
develop hand-foot syndrome due to a build-up of the toxic byproducts FBAL and
dhFU.
As these byproducts are barely detectable in patients treated with NUC-3373,
it is likely
that fewer, if any, patients treated with NUC-3373 will suffer from hand-foot
syndrome, or
any other side effect mediated by these byproducts. Indeed, in the clinical
trial disclosed in
the Examples, none of the 36 patients treated have, so far, developed hand-
foot
syndrome. As hand-foot syndrome arises in 40-60% of patients treated with 5FU,
if
clinically meaningful levels of these by products were produced following
administration of
.. NUC-3373, it would have been expected that some patients would have
developed hand-
foot syndrome. This means that NUC-3373 is likely to be the suitable treatment
choice for
cancer patients who suffer from or are likely to develop hand-foot syndrome,
or other
FBAL- or dhFU-mediated side effects. For example, a patient being treated with
5FU who
develops hand-foot syndrome could be switched from 5FU to NUC-3373 treatment;
similarly, patients that are predicted to develop hand-foot syndrome if
treated with a
particular chemotherapeutic agent (e.g. 5FU or capecitabine) could be treated
with NUC-
3373 rather than the agent that is likely to result in the development of hand-
foot
syndrome. Patients that are more likely to develop hand-foot syndrome may be
identified
based on detection of one or more suitable surrogate markers (e.g. biomarkers)
that
detect susceptibility to developing hand-foot syndrome when treated with a
chemotherapeutic drug, such as 5FU.
[0044] Further details of the various aspects and embodiments of the invention
are
described below. Technical considerations described in relation to any of the
aspects or
embodiments of the invention should also be taken as applicable to the other
aspects and
embodiments described herein, unless the context require otherwise.

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
12
[0045] The Compound
[0046] The 5-fluoro-2'-deoxyuridine-5'-041-naphthyl (benzoxy-L-alaninyI)]
phosphate
(NUC-3373) may be a mixture of phosphate diastereoisomers or it may be present
as the
(S)-epimer in substantially diastereomerically pure form or as the (R)-epimer
in
substantially diastereomerically pure form.
[0047] It may be that the NUC-3373 is not in the form of a salt. Preferably,
the NUC-
3373 is in the form of the free base.
[0048] 'Substantially diastereomerically pure' is defined for the purposes of
this
invention as a diastereomeric purity of greater than about 90% (about in this
context
means +1- 5%). If present as a substantially diastereoisomerically pure form,
the NUC-
3373 may have a diastereoisomeric purity of greater than 95%, 98%, 99%, or
even
99.5%.
[0049] Cancer to be treated
[0050] The cancer may be a cancer selected from: pancreatic cancer, breast
cancer,
ovarian cancer, bladder cancer, other urothelial cancers, gastrointestinal
cancer (also
known as cancer of the digestive tract), liver cancer, lung cancer, biliary
cancer, prostate
cancer, cholangiocarcinoma, renal cancer, neuroendocrine cancer, sarcoma,
lymphoma,
leukemia, cervical cancer, thymic cancer, a cancer of an unknown primary
origin,
mesothelioma, adrenal cancer, cancer of the uterus, cancer of the fallopian
tube,
peritoneal cancer, endometrial cancer, testicular cancer, head and neck
cancer, the
central nervous system cancer, basal cell carcinoma, Bowens disease, other
skin cancers
(such as malignant melanoma, merckel cell tumour and rare appendage tumours),
ocular
surface squamous neoplasia and germ cell tumours.
[0051] The cancer may be selected from the group consisting of: leukaemia,
lymphoma,
pancreatic cancer, prostate cancer, lung cancer, breast cancer, cervical
cancer, head and
neck cancer, ovarian cancer, and gastrointestinal cancers. The
gastrointestinal cancer
may be selected from the group consisting of: oesophageal cancer, gastric
cancer,
stomach cancer, bowel cancer, small intestine cancer, colon cancer, appendix
mucinous,
goblet cell carcinoid, liver cancer, biliary cancer, gallbladder cancer, anal
cancer and
rectal cancer.
[0052] The cancer may be relapsed. The cancer may be metastatic. The cancer
may
be previously untreated. The cancer may be refractory cancer that has
previously been
treated but has proven unresponsive to prior treatment. Alternatively, the
cancer patient
may be intolerant of a previous therapy, for example, may develop side effects
that make

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
13
the patient intolerant to further treatment with the agent being administered.
An example
of this is the development of hand-foot syndrome when receiving certain anti-
cancer
therapies, like 5FU and capecitabine or tegafur.
[0053] Suitably, treatment in accordance with the medical uses or methods of
the
invention may be provided as a first line cancer therapy (i.e. the first
cancer therapy
provided after diagnosis of the disease). Alternatively, it may be used as a
second or
further line cancer treatment. It may be used as a third line or further
cancer treatment.
[0054] Treatment regime
[0055] The first, second and third aspects of the invention all relate to
treatments
requiring the administration of NUC-3373 over a period of up to 10 hours. The
skilled
reader will appreciate that, except for where the context requires otherwise,
any of the
medical uses and methods of treatment of the invention described herein (thus,
including
those of the seventh, eighth, ninth, twelfth, and thirteenth aspects of the
invention) may
employ administration of NUC-3373 over a period of up to 10 hours.
[0056] Suitably the NUC-3373 is for use in the treatment of cancer, where
treatment is
by administration of NUC-3373 over a period of up to 9 hours, up to 8 hours,
up to 7
hours, up to 6 hours, or by administration of NUC-3373 over a period of up to
5 hours.
[0057] The NUC-3373 may be for use in the treatment of cancer, where treatment
is by
administration of NUC-3373 over a period of up to 4.75 hours, up to 4.5 hours,
up to 4
hours, up to 3.75 hours, up to 3.5 hours, up to 3.25 hours, up to 3 hours, up
to 2.75 hours,
up to 2.5 hours, up to 2.25 hours, up to 2.25 hours, up to 2 hours, up to 1.75
hours, up to
1.5 hours, up to 1.25 hours, up to 1 hour, up to 0.75 hours, up to 0.5 hours,
or by
administration over a period of up to 0.25 hours.
[0058] Suitably the NUC-3373 is for administration over a period of between 1
and 6
hours, between 1 and 5 hours, between 1 and 4 hours, between 1 and 3 hours,
between 2
and 4 hours, between 3 and 6 hours, between 3 and 5 hours or of between 1 and
2 hours.
[0059] Similarly, in methods of treatment in accordance with the various
aspects of the
invention, the NUC-3373 is suitably administered over a period of up to 9
hours, up to 8
hours, up to 7 hours, up to 6 hours, or up to 5 hours for the treatment of
cancer.
[0060] For example, the NUC-3373 may be administered over a period of up to
4.75
hours, up to 4.5 hours, up to 4 hours, up to 3.75 hours, up to 3.5 hours, up
to 3.25 hours,
up to 3 hours, up to 2.75 hours, up to 2.5 hours, up to 2.25 hours, up to 2.25
hours, up to

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
14
2 hours, up to 1.75 hours, up to 1.5 hours, up to 1.25 hours, up to 1 hour, up
to 0.75
hours, or up to 0.5 hours for the treatment of cancer.
NUC-3373 administration
[0061] Preferably, the administration is by means of infusion but could also
be by, or
include, a bolus administration.
[0062] The NUC-3373 may be administered parenterally, e.g. for intravenously,
subcutaneously or intramuscularly.
Preferably, the NUC-3373 is administered
intravenously, for example, via a central or peripheral line.
[0063] The NUC-3373 may be administered parenterally as an aqueous formulation
which optionally also comprises a polar organic solvent, e.g. DMA together
with a
surfactant. In the case of parenteral (e.g. intravenous) administration, the
formulation
preferably also comprises a polar aprotic organic solvent, e.g. DMA.
[0064] The formulation may be for dilution by a predetermined amount shortly
before
administration, i.e. up to 48 hours (e.g. up to 24, 12 or 2 hours) before
administration.
[0065] The formulation may also comprise one or more pharmaceutically
acceptable
solubilizers, e.g. a pharmaceutically acceptable non-ionic solubilizers.
Solubilizers may
also be called surfactants or emulsifiers. Illustrative solubilizers include
polyethoxylated
fatty acids and fatty acid esters and mixtures thereof. Suitable solubilizers
may be or
comprise polyethoxylated castor oil (e.g. that sold under the trade name
Kolliphor ELP);
or may be or comprise polyethoxylated hydroxy-stearic acid (e.g. that sold
under the trade
names Solutol or Kolliphor H515); or may be or comprise polyethoxylated
(e.g.
polyoxyethylene (20)) sorbitan monooleate, (e.g. that sold under the trade
name Tween
80).
[0066] In certain preferred embodiments, the formulation comprises more than
one
pharmaceutically acceptable solubilizer.
[0067] The formulation may also comprise an aqueous vehicle. The formulation
may be
ready to administer, in which case it will typically comprise an aqueous
vehicle.
[0068] The formulation may be for parenteral, e.g. for intravenous,
subcutaneous or
intramuscular administration. Preferably, the formulation is for intravenous
administration.
The administration may be through a central vein or it may be through a
peripheral vein.
[0069] The formulation may be a formulation described in W02017/109491.

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
[0070] While NUC-3373 is preferably formulated for parenteral administration,
in certain
embodiments of the invention it may be administered orally or topically.
[0071] For the above-mentioned uses and methods of the invention the dosage
administered will, of course, vary with the precise mode of administration,
the treatment
5 desired and the disorder indicated. Dosage levels, dose frequency, and
treatment
durations of compounds of the invention are expected to differ depending on
the
formulation and clinical indication, age, and co-morbid medical conditions of
the patient.
The size of the dose for therapeutic purposes of compounds of the invention
will naturally
vary according to the nature and severity of the conditions, the age and sex
of the animal
10 or patient and the route of administration, according to well known
principles of medicine.
[0072] The NUC-3373 may be administered in a dose in the range of from 100
mg/m2 to
4000 mg/m2, such as from 100 mg/m2 to 3000 mg/m2. The NUC-3373 may be
administered in a dose in the range of from 500 mg/m2 to 2000 mg/m2. The NUC-
3373
may be administered in a dose in the range of from 2000 mg/m2t0 4000 mg/m2
15 [0073] The NUC-3373 may be administered on day 1 of a 28 day cycle. It
may be
administered on days 1, 8, 15 and 22 of a 28 day cycle. It may be administered
on days 1
and 15 of a 28 day cycle.
It will be appreciated that a "cycle" is a course of treatment (treatment
cycle), typically that
is interspersed with periods of rest (no treatment).
The NUC-3373 may be administered as part of a 4, 5, 6, 7 or more series of
cycles. A
series of cycles refers to a number of sequential cycles, typically
interspersed with a
period or rest (treatment vacation).
Methods of assessing effectiveness of anti-cancer therapy
[0074] The fourth, fifth, and sixth aspects of the invention provide methods
of assessing
effectiveness of an anti-cancer therapy. The methods of the third, fourth and
fifth aspects
of the invention are of particular utility in assessing the effectiveness of
anti-cancer
therapies in subjects receiving anti-cancer treatment using NUC-3373. For
example, the
methods of the third, fourth and fifth aspects of the invention may be
utilised in respect of
a subject receiving treatment with NUC-3373 used in a medical use of the
invention (for
example a medical use of the first, third, seventh, eighth, or thirteenth
aspects of the
invention), or receiving NUC-3373 in a method of treatment in accordance with
the
invention (for example a method of treatment of the second, ninth or twelfth
aspects of the
invention).

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
16
[0075] Suitably, the level of intracellular dTMP, dUMP or TS within the PBMCs
or cancer
cells may be compared to a suitable control value. Merely by way of example, a
suitable
control value may be representative of an intracellular dTMP, dUMP or TS level
selected
from: cells of the same subject prior to receiving the anti-cancer therapy;
cells from an
individual not receiving cancer therapy; and cells of an individual receiving
cancer therapy
with an agent other than NUC-3373. The control cells may be PBMCs or
corresponding
cancer cells. The control PBMCs or cancer cells may be collected in the same
manner as
the PBMCs or cancer cells of the subject. Suitably, control values may be
generated
from historical averages. Suitably, control values are obtained by testing
cells from the
.. same patient before treatment starts (baseline levels).
[0076] As noted above, a reduction in intracellular dTMP or TS level in PBMCs
or cancer
cells as compared to a suitable control (e.g. baseline pre-treatment level) is
indicative of
effective treatment. In contrast, an increase in intracellular dUMP level in
PBMCs or
cancer cells is indicative of effective treatment. Suitably, the reduction of
intracellular
dTMP or TS level is a reduction of at least 25%. Indeed, the reduction of
intracellular
dTMP level may be a reduction of at least 50%, at least 55%, at least 60%, at
least 65%,
at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at
least 95%.
[0077] In a suitable embodiment, the reduction of intracellular dTMP or TS
level is a
substantially complete reduction of intracellular dTMP or TS. For the purposes
of the
present disclosure, a reduction of intracellular dTMP or TS level of at least
95%, at least
96%, at least 97%, at least 98%, or at least 99% may be considered a
substantially
complete reduction of intracellular dTMP or TS.
[0078] NUC-3373 works by inhibiting intracellular TS. In a suitable
embodiment, the
inhibition of intracellular TS level is substantial inhibition of
intracellular TS function. For
the purposes of the present disclosure, a inhibition of intracellular TS of at
least 95%, at
least 96%, at least 97%, at least 98%, or at least 99% may be considered a
substantially
complete inhibition of intracellular TS.
[0079] An increase in intracellular dUMP indicative of effective cancer
treatment may be
an increase of at least 25%. Suitably the increase may be an increase of at
least 50%, at
least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least
80%, at least
85%, at least 90%, or at least 95%. Indeed, a suitable increase in
intracellular dUMP may
be an increase of 100% or more.
Intracellular dTMP, TS or dUMP levels may be determined at a time
approximately in the
range from 1 to 6 hours after the subject has begun anti-cancer treatment.
Suitably,

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
17
intracellular dTMP, TS or dUMP levels may be determined at a time
approximately in the
range from 1 to 6 hours after administration of an anti-cancer agent to the
subject.
It will be appreciated that for the purposes of the present invention, the
term
"approximately" when referring to the time point for determining a biomarker,
means plus
or minus 1 hour. Suitably, intracellular dTMP or TS levels may be determined
at any time
during a treatment cycle. It will be appreciated that the relevant time is the
time of
sampling, rather than the time when the assay is carried out. For example, if
a sample is
taken after 6 hours, but frozen or treated in another way to ensure that the
amount of a
biomarker remains at roughly the same level, and the sample is then assayed
after 12
hours it is the 6 hour sampling time rather than the 12 hour assaying time
which is
relevant.
Intracellular dTMP, TS or dUMP levels may be determined by any suitable assay
or
method known to the skilled person. A suitable assay for the determination of
dTMP may
include ultra-performance liquid chromatography- tandem mass spectrometry
(UPLC-MS),
as disclosed in the Examples below, or high performance liquid chromatography
mass
spectrometry (HPLC/MS). The same technique may be used for the assessment of
intracellular dUMP levels.
Suitable methods or assays to determine TS levels may include Western blot,
immunoassays, amino acid assays, or SDS-PAGE. Detection of TS levels by
Western blot
is disclosed in the Examples below.
For example, other suitable methods for determining levels of dUMP would also
be known
to the skilled person.
In the event that a subject exhibits minimal or no reduction in intracellular
dTMP or TS, or
minimal or no increase in intracellular dUMP, a treating physician may
increase or
terminate the dose of NUC-3373 received by the subject. It will be appreciated
therefore
that levels of dTMP, TS or dUMP may be used to inform the appropriate
treatment for an
individual patient.
Dihydropyrimidine dehydrogenase (DPD) deficiency
The eighth to thirteenth aspects of the invention ail relate to medical uses
and methods
wherein a subject is deficient or partially deficient in DPD.
DPD deficiency is an autosomal recessive metabolic disorder in which there is
absent or
significantly decreased activity of DPD, an enzyme involved in the metabolism
of uracil

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
18
and thymine. The decrease in activity may result from reduced expression of
DPD, or
expression of DPD with reduced function. DPD deficiency may be manifest as
full
deficiency or partial deficiency.
[0080] Standard techniques of determining whether a cell, such as a liver
cell, PBMC or
cancer cell, is deficient in DPD are well known and the skilled person would
have the
relevant knowledge to undertake the appropriate tests. Examples of such tests
include
those set out below.
[0081] Enzymatic activity of DPD in subjects with suspected DPD deficiency can
be
determined by assaying DPD protein extracted from liver cells, PBMCs or cancer
cells.
DPD activity may also be assessed by assaying for a surrogate of DPD protein
such as
RNA extracted from liver cells, PBMCs or cancer cells. Measurement of DPD mRNA
copy
number may then be undertaken. Nucleic acids encoding DPD may also be assayed
with
reference to presence or absence of DPD gene amplification, or presence or
absence of
DPD mutations indicative of DPD, or activity of DPD in suitable cells (such as
liver cells,
PBMCs or cancer cells) extracted from the subject of interest.
[0082] The enzyme DPD is encoded by the DPYD gene in humans. It is known that
there are more than 50 mutations in the DPYD gene identified in people with
DPD
deficiency (Diagnostic Molecular Pathology: A Guide to Applied Molecular
Testing, 2017,
Edited by: William B. Coleman and Gregory J. Tsongalis).
[0083] Merely by way of example, a subject may have a genetic mutation
selected from
IV514+1G>A mutation in intron 14 coupled with exon 14 deletion (known as
DPYD*2A),
496A>G in exon 6; 2846A>T in exon 22; and T1679G (DPYD*13) in exon 13. Genetic
variants in the DPYD gene on chromosome 1p21.3 have also been shown to result
in
deficient DPD activity (Diagnostic Molecular Pathology: A Guide to Applied
Molecular
Testing, 2017, Edited by: VVilliam B. Coleman and Gregory J. Tsongalis).
[0084] In an embodiment of the fifth, sixth, seventh, eighth, ninth or tenth
aspect of the
invention, the subject has a mutation in the DPYD gene that results in
deficiency or partial
deficiency of DPD.
[0085] In another embodiment of the fifth, sixth, seventh, eighth, ninth or
tenth aspect of
the invention, the genetic mutation in a subject that results in has a
mutation in deficiency
or partial deficiency of DPD may be selected from: IV514+1G>A mutation in
intron 14
coupled with exon 14 deletion (known as DPYD*2A), 496A>G in exon 6; 2846A>T in
exon
22; and T1679G (DPYD*13) in exon 13.In another embodiment of the fifth, sixth,
seventh,

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
19
eighth, ninth or tenth aspect of the invention a subject has a mutation in
chromosome
1p21.3 of the DPYD gene that results in deficiency or partial deficiency of
DPD.
[0086] Testing for the IVS14+1G>A DPYD variant (DPYD*2A) is available
(Terrazzino,S.
et. a!, Pharmacogenomics. 2013 Aug;14(11):1255-72).
[0087] High-throughput genetic analysis using denaturing high-performance
liquid
chromatography (DH PLC) can be used, particularly if the subject is severely
neutropenic.
[0088] A subject may also be characterised as having DPD deficiency, or
partial DPD
deficiency, if they exhibit clinical or physiological characteristics of such
a deficiency.
[0089] Merely by way of example, the subject may have previously exhibited
intolerance
for 5FU or capecitabine. Such intolerance is a known clinical characteristic
of subjects
with DPD deficiency or partial deficiency. Thus, in a suitable embodiment of
the fifth,
sixth, seventh, eighth, ninth or tenth aspect of the invention the subject has
previously
exhibited intolerance for 5FU or capecitabine.
[0090] Alternatively, or additionally, the subject may have a family history
of intolerance
for 5FU or capecitabine. Thus, in a suitable embodiment, a patient selected
for use of
NUC-3373 according to the fifth, sixth, seventh, eighth, ninth or tenth aspect
of the
invention has a family history of intolerance for 5FU or capecitabine.
A known physiological characteristic of subjects with DPD deficiency or
partial deficiency
is a change in the ratio of dihydrouracil to uracil ratio in plasma. A
reduction in this ratio is
indicative of DPD deficiency or partial deficiency. Thus, in a suitable
embodiment, a
subject may be identified as having DPD deficiency or partial deficiency by
analysis of a
plasma sample to assess the ratio of dihydrouracil to uracil, and comparison
of this ratio to
suitable reference values
In a particular embodiment, the patient's cancer may be determined to be DPD
deficient
or partially deficient by testing a cancer cell or PBMC containing sample that
was
previously isolated from the patient, for the presence of DPD protein,
activity of DPD
protein, or surrogate thereof (e.g. mRNA). In a particular embodiment, the
patient's cancer
may be determined to be DPD deficient or partially deficient by testing a
cancer cell or
PBMC containing sample for the presence of genetic mutations indicative of DPD
deficiency or partial deficiency.
[0091] In an embodiment of the first and second aspects of the invention, NUC-
3373
may be administered over a period of up to 10 hours to subjects deficient or
partially
deficient in DPD. The subject may have been identified as deficient or
partially deficient in

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
DPD and selected for treatment on this basis. DPD deficiency or partial
deficiency may
have been identified by means of any of the methods disclosed here.
[0092] In another embodiment of the first and second aspects of the invention,
NUC-
3373 may be administered over a period of up to 10 hours to subjects that have
previously
5 exhibited intolerance for 5FU or capecitabine. Suitably, the subjects may
have a family
history of intolerance for 5FU or capecitabine. The subject may have been
selected for
treatment on the basis of their intolerance for 5FU or capecitabine.
A subject may be determined to have DPD deficiency or partial deficiency if
their DPD
expression or function is determined to be reduced by at least 10% (as
compared to a
10 suitable reference value) as assessed by any of the relevant tests set
out herein. Suitably
a subject with DPD deficiency or partial deficiency may have a reduction in
DPD
expression or function of at least 25%, at least 30%, least 40%, at least 50%,
least 60%,
at least 70%, least 80%, or at least 90% as compared to a suitable reference
value.
Suitably a subject with DPD deficiency may have substantially no DPD
expression or
15 function as assessed by any of the relevant tests set out herein.
Hand-foot syndrome
The fourteenth to seventeenth aspects of the invention all relate to medical
uses and
methods wherein a cancer patient/subject has or is likely to develop hand-foot
syndrome
when treated with a chemotherapeutic agent such as 5FU or capecitabine.
20 Hand-foot syndrome develops in 30-60% of patients treated with 5FU and
5FU related
fluoropyrimidines (Kruger et al. Acta Oncologica 1-8, 2015; Chiara et al. Eur
J Cancer.
33:967-969, 1997). It arises due to the build up of the 5FU toxic metabolites
dhFU and
FBAL. Hand-foot syndrome is a potentially dose -limiting cutaneous toxicity.
It is
characterized by paresthesia in a sock-and-glove distribution, µ,,vith vanying
degrees of
.. pain, tingling, dryness, erythema, scaling, swelling, and vesiculation of
the hands and feet.
Painful red swelling of the hands and feet in a patient receiving chemotherapy
is usually
enough to make the diagnosis of H&F syndrome.
In an ernbodiment, a cancer patient with hand-foot syndrome is selected for
and treated
with NUC-3373. In an embodiment, a cancer patient being treated with 5FU or
capecitabine that develops hand-foot syndrome is treated with NUC-3373;
typically, this
will mean switching the patent from 5FU or capecitabine to NUC-3373, but it
may also
involve cornbination treatment wherein the patient is administered NUC-3373 in
addition
to the 5FU.

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
21
Rather than wait to switch treatment to NUC-3373 from an agent (e.g.
therapeutic drug
like 5FU) that results in the patient developing hand-foot syndrome, it should
be possible
to use appropriate genetic analyses (e.g. DPD genetics) to predict whether any
particular
patient is likely to develop hand-foot syndrome if administered 5FU or another
chemotherapeutic agent that is known to be associated with hand-foot syndrome.
In a
particular embodiment, the patient is identified as possessing hand-foot
syndrome based
on physical examination (e.g. painful red swelling of the hands and feet in a
patient receiving
chemotherapy)
In one embodiment, following the treatment switch to NUC-3373 the hand-foot
symptoms
diminish. In a particular embodiment, the symptoms diminish entirely.
SAMPLES
[0093] It will be appreciated that the methods of the fourth, fifth, sixth,
tenth and eleventh
aspects of the invention make use of a "sample", such as PBMCs or cancer
cells. As used
herein, the term "sample" typically refers to a biological sample obtained or
derived from a
subject requiring or undergoing treatment for cancer.
[0094] In some embodiments, a biological sample consists of or comprises
biological
tissue or fluid. As set out previously, a suitable sample may comprise cancer
cells, liver
cells and/or PBMCs. In some embodiments, a biological sample may be or
comprise
blood; blood cells; plasma; bone marrow ascites; tissue or fine needle biopsy
samples;
cell-containing body fluids; free floating nucleic acids; cell free
circulating tumour DNA;
sputum; saliva; urine; cerebrospinal fluid, peritoneal fluid; pleural fluid;
faeces; lymph;
gynaecological fluids; skin swabs; vaginal swabs; oral swabs; nasal swabs;
washings or
lavages such as a ductal lavages or broncheoalveolar lavages; aspirates;
scrapings;
secretions, and/or excretions; and/or cells therefrom.
[0095] In some embodiments, a biological sample is or comprises cells obtained
from an
individual. In some embodiments, obtained cells are or include cells from a
subject. In
some embodiments, a sample is a "primary sample" obtained directly from a
source of
interest by any appropriate means. For example, in some embodiments, a primary
biological sample is obtained by methods selected from the group consisting of
biopsy
(e.g., fine needle aspiration or tissue biopsy), surgery, collection of body
fluid (e.g., blood,
lymph, ascites, faeces).
[0096] In some embodiments, as will be clear from context, the term "sample"
refers to a
preparation that is obtained by processing (e.g., by removing one or more
components of
and/or by adding one or more agents to) a primary sample. For example,
filtering using a

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
22
semi-permeable membrane. Such a "processed sample" may comprise, for example
nucleic acids or proteins extracted from a sample or obtained by subjecting a
primary
sample to techniques such as amplification (e.g. polymerase chain reaction) or
reverse
transcription of mRNA, isolation and/or purification of certain components.
[0097] A suitable sample may be selected on the basis of its ability to
contain an analyte
to be analysed, such as DPD protein (or a surrogate thereof), dTMP, dUMP, TS,
dhFU, or
FBAL.
[0098] In some embodiments, the sample maybe a liquid, solid, or mixed
biological
sample obtained from a subject having, or suspected of having, a DPD deficient
cancer.
Suitable tissue samples include cancer tissue samples including those that may
be
obtained by a biopsy or following surgical resection of the cancer,
surrounding tissues,
and/or distant tissues in which metastasis are known or are suspected.
[0099] The diagnostic/determining methods of the invention can be undertaken
using a
sample previously taken from the individual or patient. Such samples may be
preserved
by freezing or fixed and embedded in formalin-paraffin or other media.
Alternatively, a
fresh cancer cell containing sample may be obtained and used directly or
frozen and
tested later.
[00100] As noted above, the presence of DPD protein can be detected in the
cells,
including the cell nuclei, using any of a variety of techniques. In particular
embodiments,
the presence of DPD protein is detected using immunohistochemistry,
immunofluorescence, Western blotting, capillary electrophoresis, flow-
cytometry or ELISA.
Furthermore, these methods can be employed using an antibody or digital
barcoded
antibody to DPD protein. A digital barcoded antibody is an antibody whereby
DNA
barcodes are attached to the antibody. Multiple barcoded antibodies can then
be assayed
in parallel and subsequently analysed by DNA sequencing (e.g. see Agasti et
al. J Am
Chem Soc. 134(45):18499-18502,2012)
[00101] In general, the level of DPD can be assessed using any of a variety of
methods.
In many embodiments, the level of DPD expression is assessed by determining
the level
of an DPD gene product in a sample obtained from a tumour. DPD protein level
can also
be determined using a surrogate of DPD protein, such as for example mRNA
encoding
DPD. Optionally the mRNA is detected directly or measured after conversion to
cDNA
which may optionally be amplified (e.g. by reverse transcriptase PCR).
[00102] The skilled person will readily be able to determine suitable
reference values with
respect to which the amount of the appropriate target molecule (e.g. DPD) may
be

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
23
compared. Merely by way of example, expression of target molecule in cancerous
tissue
can be compared to expression of that same molecule in non-cancerous tissue,
such as
adjacent non-cancerous tissue. Expression can be assessed on a protein level
for
example by immunohistochemistry or on a DNA level for example by fluorescence
in situ
hybridization, or on a RNA level, for example by quantitative real-time PCR.
COMPOUNDS, DOSAGES FORMULATIONS OF THE INVENTION
[00103] Throughout this specification, the term S-epimer or S-diastereoisomer
refers to 5-
fluoro-2'-deoxyuridine-5'-041-naphthyl (benzoxy-L-alaninyI)]-(S)-phosphate.
Likewise,
throughout this specification, the term R-epimer or R-diastereoisomer refers
to 5-fluoro-2'-
deoxyuridine-5'-0[l-naphthyl (benzoxy-L-alaninyI)]-(R)-phosphate.
[00104] The compounds of the invention may be obtained, stored and/or
administered in
the form of a pharmaceutically acceptable salt. Suitable pharmaceutically
acceptable
salts include, but are not limited to, salts of pharmaceutically acceptable
inorganic acids
such as hydrochloric, sulphuric, phosphoric, nitric, carbonic, boric,
sulfamic, and
hydrobromic acids, or salts of pharmaceutically acceptable organic acids such
as acetic,
propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, malic, citric,
lactic, mucic,
gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulphonic,
toluenesulphonic,
benzenesulphonic, salicylic, sulphanilic, aspartic, glutamic, edetic, stearic,
palmitic, oleic,
lauric, pantothenic, tannic, ascorbic and valeric acids. Suitable base salts
are formed
from bases which form non-toxic salts. Examples include the aluminium,
arginine,
benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine,
magnesium,
meglumine, olamine, potassium, sodium, tromethamine and zinc salts. Hemisalts
of acids
and bases may also be formed, for example, hemisulfate, hemioxalate and
hemicalcium
salts. In certain embodiments, particularly those that apply to the s-epimer,
the compound
is in the form of a HCI salt or a hemioxalate salt.
[00105] Compounds of the invention may exist in a single crystal form or in a
mixture of
crystal forms or they may be amorphous. Thus, compounds of the invention
intended for
pharmaceutical use may be administered as crystalline or amorphous products.
They may
be obtained, for example, as solid plugs, powders, or films by methods such as
precipitation, crystallization, freeze drying, or spray drying, or evaporative
drying.
Microwave or radio frequency drying may be used for this purpose.
[00106] A compound of the invention, or pharmaceutically acceptable salt
thereof, may be
used on their own but will generally be administered in the form of a
pharmaceutical
composition in which the compounds of the invention, or pharmaceutically
acceptable salt

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
24
thereof, is in association with a pharmaceutically acceptable adjuvant,
diluent or carrier.
Conventional procedures for the selection and preparation of suitable
pharmaceutical
formulations are described in, for example, "Pharmaceuticals - The Science of
Dosage
Form Designs", M. E. AuIton, Churchill Livingstone, 1988.
[00107] Depending on the mode of administration of the compounds of the
invention, the
pharmaceutical composition which is used to administer the compounds of the
invention
will preferably comprise from 0.05 to 99 %w (per cent by weight) compounds of
the
invention, more preferably from 0.05 to 80 %w compounds of the invention,
still more
preferably from 0.10 to 70 %w compounds of the invention, and even more
preferably
from 0.10 to 50 %w compounds of the invention, all percentages by weight being
based
on total composition.
[00108] For oral administration the compounds of the invention may be admixed
with an
adjuvant or a carrier, for example, lactose, saccharose, sorbitol, mannitol; a
starch, for
example, potato starch, corn starch or amylopectin; a cellulose derivative; a
binder, for
example, gelatine or polyvinylpyrrolidone; and/or a lubricant, for example,
magnesium
stearate, calcium stearate, polyethylene glycol, a wax, paraffin, and the
like, and then
compressed into tablets. If coated tablets are required, the cores, prepared
as described
above, may be coated with a concentrated sugar solution which may contain, for
example,
gum arabic, gelatine, talcum and titanium dioxide. Alternatively, the tablet
may be coated
with a suitable polymer dissolved in a readily volatile organic solvent.
[00109] For the preparation of soft gelatine capsules, the compounds of the
invention
may be admixed with, for example, a vegetable oil or polyethylene glycol. Hard
gelatine
capsules may contain granules of the compound using either the above-mentioned
excipients for tablets. Also liquid or semisolid formulations of the compound
of the
invention may be filled into hard gelatine capsules.
[00110] Liquid preparations for oral application may be in the form of syrups
or
suspensions, for example, solutions containing the compound of the invention,
the
balance being sugar and a mixture of ethanol, water, glycerol and propylene
glycol.
Optionally such liquid preparations may contain colouring agents, flavouring
agents,
sweetening agents (such as saccharine), preservative agents and/or
carboxymethylcellulose as a thickening agent or other excipients known to
those skilled in
art.
[00111] For parenteral (e.g. intravenous) administration the compounds may be
administered as a sterile aqueous or oily solution. The compounds of the
invention are

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
very lipophillic. Aqueous formulations will typically, therefore, also contain
a
pharmaceutically acceptable polar organic solvent.
[00112] The present invention also includes all pharmaceutically acceptable
isotopically-
labelled forms of NUC-3373 wherein one or more atoms are replaced by atoms
having the
5 same atomic number, but an atomic mass or mass number different from the
atomic mass
or mass number of the predominant isotope usually found in nature.
[00113] Examples of isotopes suitable for inclusion in the compounds of the
invention
include isotopes of hydrogen, such as 2H and 3H, carbon, such as 110,
130 and 140,
chlorine, such as 3601, fluorine, such as 18F, iodine, such as 1231 and 1251,
nitrogen, such as
10 13N and 15N, oxygen, such as 150, 170 and 180, phosphorus, such as 32P,
and sulphur,
such as 35S.
[00114] Certain isotopically-labelled compounds, for example, those
incorporating a
radioactive isotope, are useful in drug and/or substrate tissue distribution
studies. The
radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 140, are
particularly useful for this
15 purpose in view of their ease of incorporation and ready means of
detection.
[00115] Substitution with heavier isotopes such as deuterium, i.e. 2H, may
afford certain
therapeutic advantages resulting from greater metabolic stability, for
example, increased
in vivo half-life or reduced dosage requirements, and hence may be preferred
in some
circumstances.
20 [00116] Substitution with positron emitting isotopes, such as 110, 18F,
150 and 13N, U N, can be
useful in Positron Emission Topography (PET) studies for examining substrate
receptor
occupancy.
[00117] Isotopically-labelled compounds can generally be prepared by
conventional
techniques known to those skilled in the art or by processes analogous to
those described
25 using an appropriate isotopically-labelled reagent in place of the non-
labelled reagent
previously employed.
COMBINATIONS
[00118] The method of treatment or the compound for use in the treatment of
cancer may
involve, in addition to the NUC-3373, conventional surgery or radiotherapy or
chemotherapy. Such chemotherapy may include the administration of one or more
other
active agents.
[00119] Thus, the, each or any one of the pharmaceutical formulations may
comprise
another active agent.

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
26
[00120] The one or more other active agents may be one or more of the
following
categories of anti-tumour agents:
(i) antiproliferative/antineoplastic drugs and combinations thereof, such
as alkylating
agents (for example cyclophosphamide, nitrogen mustard, bendamustin,
melphalan,
chlorambucil, busulphan, temozolamide and nitrosoureas); antimetabolites (for
example
gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and
tegafur,
raltitrexed, methotrexate, pemetrexed, cytosine arabinoside, and hydroxyurea);
antibiotics
(for example anthracyclines like adriamycin, bleomycin, doxorubicin,
daunomycin,
epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin);
antimitotic agents (for
example vinca alkaloids like vincristine, vinblastine, vindesine and
vinorelbine and taxoids
like taxol and taxotere and polokinase inhibitors); proteasome inhibitors, for
example
carfilzomib and bortezomib; interferon therapy; and topoisomerase inhibitors
(for example
epipodophyllotoxins like etoposide and teniposide, amsacrine, topotecan,
mitoxantrone
and camptothecin);
(ii) cytostatic agents such as antiestrogens (for example tamoxifen,
fulvestrant,
toremifene, raloxifene, droloxifene and iodoxyfene), antiandrogens (for
example
bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists
or LHRH
agonists (for example goserelin, leuprorelin and buserelin), progestogens (for
example
megestrol acetate), aromatase inhibitors (for example as anastrozole,
letrozole, vorazole
and exemestane) and inhibitors of 5a-reductase such as finasteride;
(iii) anti-invasion agents, for example dasatinib and bosutinib (SKI-606), and
metalloproteinase inhibitors, inhibitors of urokinase plasminogen activator
receptor
function or antibodies to Heparanase;
(iv) inhibitors of growth factor function: for example such inhibitors include
growth factor
antibodies and growth factor receptor antibodies, for example the anti-erbB2
antibody
trastuzumab [HerceptinTm], the anti-EGFR antibody panitumumab, the anti-erbB1
antibody
cetuximab, tyrosine kinase inhibitors, for example inhibitors of the epidermal
growth factor
family (for example EGFR family tyrosine kinase inhibitors such as gefitinib,
erlotinib and
6-acrylamido-N-(3-chloro-4-fluorophenyI)-7-(3-morpholinopropoxy)-quinazolin-4-
amine (Cl
1033), erbB2 tyrosine kinase inhibitors such as lapatinib); inhibitors of the
hepatocyte
growth factor family; inhibitors of the insulin growth factor family;
modulators of protein
regulators of cell apoptosis (for example BcI-2 inhibitors); inhibitors of the
platelet-derived
growth factor family such as imatinib and/or nilotinib (AMN107); inhibitors of
serine/threonine kinases (for example Ras/Raf signalling inhibitors such as
farnesyl
transferase inhibitors, for example sorafenib , tipifarnib and lonafarnib),
inhibitors of cell

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
27
signalling through MEK and/or AKT kinases, c-kit inhibitors, abl kinase
inhibitors, PI3
kinase inhibitors, Plt3 kinase inhibitors, CSF-1R kinase inhibitors, IGF
receptor, kinase
inhibitors; aurora kinase inhibitors and cyclin dependent kinase inhibitors
such as CDK2
and/or CDK4 inhibitors;
(v) antiangiogenic agents such as those which inhibit the effects of
vascular endothelial
growth factor, [for example the anti-vascular endothelial cell growth factor
antibody
bevacizumab (AvastinTm); thalidomide; lenalidomide; and for example, a VEGF
receptor
tyrosine kinase inhibitor such as vandetanib, vatalanib, sunitinib, axitinib
and pazopanib;
(vi) gene therapy approaches, including for example approaches to replace
aberrant
genes such as aberrant p53 or aberrant BRCA1 or BRCA2;
(vii) immunotherapy approaches, including for example antibody therapy such as
alemtuzumab, rituximab, ibritumomab tiuxetan (Zevaline) and ofatumumab;
interferons
such as interferon a; interleukins such as IL-2 (aldesleukin); interleukin
inhibitors for
example IRAK4 inhibitors; cancer vaccines including prophylactic and treatment
vaccines
such as HPV vaccines, for example Gardasil, Cervarix, Oncophage and Sipuleucel-
T
(Provenge); toll-like receptor modulators for example TLR-7 or TLR-9 agonists;
checkpoint
inhibitors, such as anti-PD1, anti-PD-L1 and anti-CTLA monoclonal antibodies
such as:
nivolumab, pembrolizumab, pidilizumab, atezolizumab, durvalumab and avelumab;
(viii) cytotoxic agents for example fludaribine (fludara), cladribine,
pentostatin (Nipent-rm);
irinotecan and oxaliplatin;
(ix) steroids such as corticosteroids, including glucocorticoids and
mineralocorticoids, for
example aclometasone, aclometasone dipropionate, aldosterone, amcinonide,
beclomethasone, beclomethasone dipropionate, betamethasone, betamethasone
dipropionate, betamethasone sodium phosphate, betamethasone valerate,
budesonide,
clobetasone, clobetasone butyrate, clobetasol propionate, cloprednol,
cortisone, cortisone
acetate, cortivazol, deoxycortone, desonide, desoximetasone, dexamethasone,
dexamethasone sodium phosphate, dexamethasone isonicotinate,
difluorocortolone,
fluclorolone, flumethasone, flunisolide, fluocinolone, fluocinolone acetonide,
fluocinonide,
fluocortin butyl, fluorocortisone, fluorocortolone, fluocortolone caproate,
fluocortolone
pivalate, fluorometholone, fluprednidene, fluprednidene acetate,
flurandrenolone,
fluticasone, fluticasone propionate, halcinonide, hydrocortisone,
hydrocortisone acetate,
hydrocortisone butyrate, hydrocortisone aceponate, hydrocortisone buteprate,
hydrocortisone valerate, icomethasone, icomethasone enbutate, meprednisone,
methylprednisolone, mometasone paramethasone, mometasone furoate monohydrate,

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
28
prednicarbate, prednisolone, prednisone, tixocortol, tixocortol pivalate,
triamcinolone,
triamcinolone acetonide, triamcinolone alcohol and their respective
pharmaceutically
acceptable derivatives. A combination of steroids may be used, for example a
combination of two or more steroids mentioned in this paragraph;
(x) targeted therapies, for example PI3Kd inhibitors, for example idelalisib
and
perifosine; or compounds that inhibit PD-1, PD-L1 and CAR T; and
(xi) agents that enhance the anti-cancer effect of chemotherapeutic drugs,
e.g. leucovorin.
[00121] The one or more other active agents may also be antibiotics.
[00122] Throughout the description and claims of this specification, the words
"comprise"
and "contain" and variations of them mean "including but not limited to", and
they are not
intended to (and do not) exclude other moieties, additives, components,
integers or steps.
Throughout the description and claims of this specification, the singular
encompasses the
plural unless the context otherwise requires. In particular, where the
indefinite article is
used, the specification is to be understood as contemplating plurality as well
as
singularity, unless the context requires otherwise.
[00123] Features, integers, characteristics, compounds, chemical moieties or
groups
described in conjunction with a particular aspect, embodiment or example of
the invention
are to be understood to be applicable to any other aspect, embodiment or
example
described herein unless incompatible therewith. All of the features disclosed
in this
specification (including any accompanying claims, abstract and drawings),
and/or all of the
steps of any method or process so disclosed, may be combined in any
combination,
except combinations where at least some of such features and/or steps are
mutually
exclusive. The invention is not restricted to the details of any foregoing
embodiments.
The invention extends to any novel one, or any novel combination, of the
features
disclosed in this specification (including any accompanying claims, abstract
and
drawings), or to any novel one, or any novel combination, of the steps of any
method or
process so disclosed.
[00124] The reader's attention is directed to all papers and documents which
are filed
concurrently with or previous to this specification in connection with this
application and
which are open to public inspection with this specification, and the contents
of all such
papers and documents are incorporated herein by reference.

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
29
The invention will now be further described with reference to the following
Examples and
accompanying Figures.
Example 1 ¨ Pharmacokinetic analysis from the NuTide:301 study
NuTide:301 is a Phase 1 dose escalation study in patients with advanced solid
tumors.
All patients have metastatic spread. To date, data has been generated from 21
out of 36
patients enrolled in the study, having a median age of 57 (range 20 to 77) and
having had
an average of three (range two to five) prior chemotherapy regimens. There
have been
primary cancer types, the majority of which (57%) are colorectal cancer.
10 The patients received NUC-3373, administered as a 30-minute to 2hour
intravenous
injection on days 1, 8, 15 and 22 of a 28-day cycle regimen. Patients could
remain on
study and receive treatment until disease progression or unacceptable toxicity
occurs.
NUC-3373 is presented as a single dose intravenous injection in a clear vial
containing
250 mg/ml NUC-3373 in a solution of dimethylacetamide (DMA) and normal saline
in the
ratio of 80:20. The product is a clear yellow solution, free from visible
particles.
In the study all patients were treated with a 1:1 mixture of NUC-3373 Sand R
epimers.
The cohorts treated to date received 125 mg/m2, 250 mg/m2, 500 mg/m2 or 750
mg/m2
NUC-3373 per administration.
Pharmacokinetic analysis of blood samples of the patients was then carried
out. The
results are shown in Figures 1 and 2.
Blood samples were collected on days 1 and 15 of a treatment cycle:
The blood samples were taken at the following 12 time points listed below in
Table 1.
Table 1: Sampling Schedule
Sample 1 TO pre-dose (before the start of the infusion)
Sample 2 Ti (immediately after drug has cleared the infusion line)
Sample 3 Ti plus 15 minutes
Sample 4 Ti plus 30 minutes
Sample 5 Ti plus 45 minutes
Sample 6 Ti plus 1 hour

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
Sample 7 Ti plus 1.5 hours
Sample 8 Ti plus 2 hours
Sample 9 Ti plus 4 hours
Sample 10 Ti plus 6 hours
Sample 11 Ti plus 24 hours
Sample 12* Ti plus 48 hours
*Optional
Processing of Plasma Samples- 4m1 Lithium Heparin Vacutainer
The blood samples should arrive in the lab within 2 hours of collection.
Centrifuge the 4m1
blood sample at 1,200g at 18 C for 10 minutes. Using a sterile plastic pipette
(pastette),
5 remove the resulting plasma and transfer -1.0m1 plasma into each of 2
cryovials (2m1).
Processing of PBMC Sample- 8m1 CPT Tubes
The blood samples should arrive in the lab within 2 hours of collection.
Collect 8m1 of
blood into CPT tubes (blood tubes should be centrifuged within 2 hours of
blood
collection). Remix the blood sample immediately prior to centrifugation by
gently inverting
10 the tube 8 to 10 times. Set centrifuge so that the start and finish is
set to a slow
acceleration (without break). Centrifuge at 18 C for 20 minutes at 1,500g.
Remove
sample from the centrifuge carefully. This will result in five layers: plasma
(first); whitish
cell (PBMC) layer [second]; polyester gel [third]; density solution [fourth]
and, remaining
granulocytes and RBCs [fifth]. Aspirate approximately half of the plasma
without
15 disturbing the cell layer. Collect the PBMC layer with a Pasteur Pipette
and transfer to a
50m1 tube and add cold PBS (4 C) to a final volume of 40m1. Divide between two
50m1
tubes (20m1 per tube) and centrifuge for 5 minutes at 4 C at 1,500g. Decant
out the
supernatant without disturbing the cell pellet and re-suspend the pellets in
the residual
buffer. Add lml PBS to each pellet to gather up the cells and transfer one
pellet to a 2m1
20 screw cap tube labelled 'PD PBMCs' and the other to a 2m1 screw cap tube
labelled `PK
PBMCs Pellet'. Centrifuge both tubes for 5 minutes at 4 C at 1,500g. Remove
the
supernatant with a pipette (p 1,000 pl). Please ensure that the supernatant is
completely
removed from the tube to ensure good quality sample. Add 200 pl of freezing
media
solution (provided, 4 C) to the tube labelled 'PD PBMCs' and store at -80 C
until analysis.

CA 03073649 2020-02-21
WO 2019/043392 PCT/GB2018/052455
31
Re-suspend the cell pellets in the tube labelled `PK PBMCs Pellet' by adding
200 pl of 8-
% Methanol (4 C) and gently pipette up and down for 3 times. Vortex-mix for 30
seconds.
Leave sample on ice for 15 minutes. Centrifuge at 4 C for 5 minutes at 1,500g.
Using a
micropipette, carefully transfer 180 pl of the supernatant to a 2m1 screw cap
tube labelled
.. `PK PBMC Supernatant' without disturbing the cell pellet. Store both the
PBMC pellet and
PBMC supernatant at -80 C until analysis for dTMP, TS, dUMP, FBAL and dhFU.
UPLC-MS for was used to detect dTMP Western blot was used to detect TS.
UPLC-MS/MS was used to detect dhFU and FBAL
Figure 1 shows the Cm, and AUC for NUC-3373 in the blood plasma for the
cohorts
treated to date. The half-life of NUC-3373 in plasma was 9.7 hours. In
contrast, 5FU has
a plasma half-life of 8-14 minutes. The toxic byproducts a-fluoro-p-alanine
(FBAL) and
dihydrofluorouracil (dhFU) were undetectable following NUC-3373 administration
at the
doses studied.
Figure 2 shows the Cm, and AUC for intracellular FUDR monophosphate for the
cohorts
treated to date. The half-life of FUDR monophosphate was found to be 14.9
hours.
FUDR monophosphate was still detectable at 48 hours.
It is known that accumulation of dUMP within cells leads to DNA damage, and
that this
DNA damage is associated with cell death. Thus, the ability of NUC-3373 to
promote
accumulation of dUMP within cancer cells represents a previously unrecognized
mode of
action by which NUC-3373 is able to kill cancer cells, and thereby effectively
treat cancer.

Dessin représentatif

Désolé, le dessin représentatif concernant le document de brevet no 3073649 est introuvable.

États administratifs

2024-08-01 : Dans le cadre de la transition vers les Brevets de nouvelle génération (BNG), la base de données sur les brevets canadiens (BDBC) contient désormais un Historique d'événement plus détaillé, qui reproduit le Journal des événements de notre nouvelle solution interne.

Veuillez noter que les événements débutant par « Inactive : » se réfèrent à des événements qui ne sont plus utilisés dans notre nouvelle solution interne.

Pour une meilleure compréhension de l'état de la demande ou brevet qui figure sur cette page, la rubrique Mise en garde , et les descriptions de Brevet , Historique d'événement , Taxes périodiques et Historique des paiements devraient être consultées.

Historique d'événement

Description Date
Rapport d'examen 2024-09-16
Paiement d'une taxe pour le maintien en état jugé conforme 2024-08-02
Requête visant le maintien en état reçue 2024-07-31
Lettre envoyée 2023-09-27
Requête d'examen reçue 2023-08-25
Exigences pour une requête d'examen - jugée conforme 2023-08-25
Modification reçue - modification volontaire 2023-08-25
Toutes les exigences pour l'examen - jugée conforme 2023-08-25
Modification reçue - modification volontaire 2023-08-25
Représentant commun nommé 2020-11-07
Inactive : Page couverture publiée 2020-04-16
Inactive : CIB attribuée 2020-03-25
Inactive : CIB attribuée 2020-03-25
Inactive : CIB attribuée 2020-03-25
Inactive : CIB attribuée 2020-03-20
Inactive : CIB enlevée 2020-03-20
Inactive : CIB en 1re position 2020-03-20
Inactive : CIB enlevée 2020-03-20
Inactive : CIB attribuée 2020-03-20
Lettre envoyée 2020-02-28
Demande reçue - PCT 2020-02-27
Inactive : CIB en 1re position 2020-02-27
Inactive : CIB attribuée 2020-02-27
Inactive : CIB attribuée 2020-02-27
Inactive : CIB attribuée 2020-02-27
Inactive : CIB attribuée 2020-02-27
Demande de priorité reçue 2020-02-27
Exigences applicables à la revendication de priorité - jugée conforme 2020-02-27
Exigences pour l'entrée dans la phase nationale - jugée conforme 2020-02-21
Demande publiée (accessible au public) 2019-03-07

Historique d'abandonnement

Il n'y a pas d'historique d'abandonnement

Taxes périodiques

Le dernier paiement a été reçu le 2024-07-31

Avis : Si le paiement en totalité n'a pas été reçu au plus tard à la date indiquée, une taxe supplémentaire peut être imposée, soit une des taxes suivantes :

  • taxe de rétablissement ;
  • taxe pour paiement en souffrance ; ou
  • taxe additionnelle pour le renversement d'une péremption réputée.

Les taxes sur les brevets sont ajustées au 1er janvier de chaque année. Les montants ci-dessus sont les montants actuels s'ils sont reçus au plus tard le 31 décembre de l'année en cours.
Veuillez vous référer à la page web des taxes sur les brevets de l'OPIC pour voir tous les montants actuels des taxes.

Historique des taxes

Type de taxes Anniversaire Échéance Date payée
TM (demande, 2e anniv.) - générale 02 2020-08-31 2020-02-21
Taxe nationale de base - générale 2020-02-21 2020-02-21
TM (demande, 3e anniv.) - générale 03 2021-08-30 2021-08-02
TM (demande, 4e anniv.) - générale 04 2022-08-30 2022-08-01
TM (demande, 5e anniv.) - générale 05 2023-08-30 2023-08-22
Requête d'examen - générale 2023-08-30 2023-08-25
TM (demande, 6e anniv.) - générale 06 2024-08-30 2024-07-31
Titulaires au dossier

Les titulaires actuels et antérieures au dossier sont affichés en ordre alphabétique.

Titulaires actuels au dossier
NUCANA PLC
Titulaires antérieures au dossier
HUGH GRIFFITH
Les propriétaires antérieurs qui ne figurent pas dans la liste des « Propriétaires au dossier » apparaîtront dans d'autres documents au dossier.
Documents

Pour visionner les fichiers sélectionnés, entrer le code reCAPTCHA :



Pour visualiser une image, cliquer sur un lien dans la colonne description du document. Pour télécharger l'image (les images), cliquer l'une ou plusieurs cases à cocher dans la première colonne et ensuite cliquer sur le bouton "Télécharger sélection en format PDF (archive Zip)" ou le bouton "Télécharger sélection (en un fichier PDF fusionné)".

Liste des documents de brevet publiés et non publiés sur la BDBC .

Si vous avez des difficultés à accéder au contenu, veuillez communiquer avec le Centre de services à la clientèle au 1-866-997-1936, ou envoyer un courriel au Centre de service à la clientèle de l'OPIC.


Description du
Document 
Date
(aaaa-mm-jj) 
Nombre de pages   Taille de l'image (Ko) 
Revendications 2023-08-24 3 157
Description 2020-02-20 31 1 701
Revendications 2020-02-20 4 193
Abrégé 2020-02-20 1 49
Dessins 2020-02-20 2 81
Demande de l'examinateur 2024-09-15 7 166
Confirmation de soumission électronique 2024-07-30 2 65
Courtoisie - Lettre confirmant l'entrée en phase nationale en vertu du PCT 2020-02-27 1 586
Courtoisie - Réception de la requête d'examen 2023-09-26 1 422
Modification / réponse à un rapport / Requête d'examen 2023-08-24 13 612
Rapport de recherche internationale 2020-02-20 5 159
Demande d'entrée en phase nationale 2020-02-20 8 202